scholarly journals 379 Maternal Plasma Urocortin I Levels in Preeclampsia and Fetal Growth Restriction Predict Neonatal Intraventricular Haemorrhage

2005 ◽  
Vol 58 (2) ◽  
pp. 419-419
Author(s):  
M Torricelli ◽  
P Florio ◽  
A Giovannelli ◽  
P B Torres ◽  
A Dell'Anna ◽  
...  
Author(s):  
Mujde Can Ibano ◽  
Cem Yasar Sanhal ◽  
Seval Ozgu-Erdinc ◽  
Aykan Yucel

Background: Higher Fetuin-A (FA) concentrations were found to be associated with obesity and there is an interest to the relation between maternal FA and pregnancy outcomes. Objective: In this study, our aim was to evaluate the association of maternal plasma levels of FA with fetal growth restriction (FGR). Materials and Methods: 41 pregnant women with FGR and 40 controls were recruited in this case-control study between July and November 2015. At the diagnosis of FGR, venous blood samples (10 cc) were obtained for FA analysis. Results: Maternal plasma FA levels were significantly higher in fetal growth-restricted pregnant women compared with controls (19.3 ± 3.0 ng/ml vs 25.9 ± 6.8 ng/ml, p = 0.001). Area under receiver operating characteristic curve analysis of FA in FGR was 0.815 (95% confidence interval (CI): 0.718-0.912, p < 0.001). The maternal FA levels with values more than 22.5 ng/ml had a sensitivity of about 73.17% (95% CI: 56.79- 85.25) and a specificity of about 82.5% (95% CI: 66.64-92.11) with positive and negative predictive values of about 81.08% (95% CI: 64.29-91.45) and 75% (95% CI: 59.35-86.30), respectively. Therefore, the diagnostic accuracy was obtained about 77.78%. Conclusion: The results of this study show higher maternal plasma levels of FA in FGR. Further studies are needed in order to demonstrate the long-term effects of FA in pregnancies complicated with FGR and early prediction of FGR.


2006 ◽  
Vol 2 (3-4) ◽  
pp. 169-184 ◽  
Author(s):  
Madhulika B. Gupta ◽  
Maxim D. Seferovic ◽  
Suya Liu ◽  
Robert J. Gratton ◽  
Amanda Doherty-Kirby ◽  
...  

2021 ◽  
Vol 12 ◽  
Author(s):  
Huan Yang ◽  
Jie He ◽  
Shuai Huang ◽  
Hongbing Yang ◽  
Qingjie Yi ◽  
...  

Many studies have confirmed that extrachromosomal circular DNAs (eccDNAs/ecDNAs) exist in tumor and normal cells independently of the chromosome and are essential for oncogene plasticity and drug resistance. Studies have confirmed that there are many eccDNAs/ecDNAs in maternal plasma derived from the fetus. Fetal growth restriction (FGR) is a pregnancy-related disease associated with high newborn morbidity and mortality. However, the characteristics and nature of eccDNAs/ecDNAs in FGR are poorly understood. This study aims to deconstruct the properties and potential functions of eccDNAs/ecDNAs in FGR. We performed circle-seq to identify the expression profile of eccDNAs/ecDNAs, analyzed by bioinformatics, and verified by real-time Polymerase Chain Reaction (PCR) combined with southern blot in FGR compared with the normal groups. A total of 45,131 eccDNAs/ecDNAs (including 2,118 unique ones) were identified, which had significantly higher abundance in FRG group than in normal group, and was bimodal in length, peaking at ~146bp and ~340bp, respectively. Gestational age may be one independent factor affecting the production of eccDNAs/ecDNAs, most of which come from genomic regions with high gene density, with a 4~12bp repeat around the junction, and their origin had a certain genetic preference. In addition, some of the host-genes overlapped with non-coding RNAs (ncRNAs) partially or even completely. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that host-genes on the differentially expressed eccDNAs/ecDNAs (DEEECs/DEECs) were mainly enriched in immune-related functions and pathways. The presence of some ecDNAs were verified, and whose variability were consistent with the circle-seq results. We identified and characterized eccDNAs/ecDNAs in placentas with FGR, and elucidated the formation mechanisms and the networks with ncRNAs, which provide a new vision for the screening of new biomarkers and therapeutic targets for FGR.


Hypertension ◽  
2006 ◽  
Vol 47 (2) ◽  
pp. 203-208 ◽  
Author(s):  
Sofía P. Salas ◽  
Guillermo Marshall ◽  
Blanca L. Gutiérrez ◽  
Pedro Rosso

2016 ◽  
Vol 30 (6) ◽  
pp. 635-639 ◽  
Author(s):  
Kirsten R. Palmer ◽  
Tu’uhevaha J. Kaitu’u-Lino ◽  
Ping Cannon ◽  
Laura Tuohey ◽  
Manarangi S. De Silva ◽  
...  

2021 ◽  
Vol 6_2021 ◽  
pp. 60-65
Author(s):  
Kan N.E. Kan ◽  
Tyutyunnik V.L. Tyutyunnik ◽  
Khachatryan Z.V. Khachatryan ◽  
Sadekova A.A. Sadekova ◽  
Krasnyi A.M. Krasnyi ◽  
...  

2007 ◽  
Vol 54 (6) ◽  
pp. 945-951 ◽  
Author(s):  
Hiroko MISE ◽  
Shigeo YURA ◽  
Hiroaki ITOH ◽  
Mercy A. NUAMAH ◽  
Maki TAKEMURA ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 341-341
Author(s):  
Seisuke Sayama ◽  
Anren Song ◽  
Yang Xia

Background: Insufficient oxygen supply is associated with the pathophysiology of fetal growth restriction (FGR). Although the erythrocyte (RBC) is the most abundant and only cell type to deliver oxygen, its function and regulatory mechanism in FGR remains unknown. Recently, adenosine uptake by equilibrative nucleoside transporter 1 (ENT1), a key adenosine transporter expressed in RBCs, was reported to be crucial for RBCs to deliver oxygen. We aimed to investigate the involvement of RBCs' oxygen delivering capacity in maintaining fetal growth by focusing on RBC ENT1. Methods and Results: The mating strategy was to delete ENT1 only on the maternal RBCs but not in the placentas or fetuses to assess the effect of maternal RBC ENT1 on fetal growth. Specifically, EpoR-Cre+ (EPO) female mouse was used as a control and Ent1f/f-EpoR-Cre+ (E1FE) female mouse as an experimental mouse and mated with WT male mouse. As a result, E1FE dams showed FGR phenotype with reduction of 12.9% in fetal weight compared to EPO group. The maternal RBCs showed decrease in p50 and 2,3-BPG in E1FE, indicating decreased oxygen delivery in E1FE RBCs. To determine the molecular basis underlying the FGR phenotype seen in EIFE dams, we conducted a metabolomics screening of the RBCs isolated from controls and EIFE dams. It showed that adenosine metabolism inside the RBCs is the most impacted pathway. Specifically, it showed decrease in adenosine, AMP, and hypoxanthine, but adenine, ADP, and ATP did not show any reduction, implicating that ENT1-mediated uptake of adenosine is largely converted to AMP. We then incubated either WT or ENT1 KO RBCs with isotopically 13C15N labeled adenosine and traced the metabolism of intracellular adenosine derived from labeled adenosine. Indeed, adenosine was rapidly phosphorylated to AMP upon uptake, and 13C15N labeled AMP levels were lower in the ENT1 KO RBCs compared to controls. These findings provide evidence that 1) the most affected metabolic pathway in the RBCs of EIFE dam is adenosine metabolism; 2) ENT1-mediated uptake of extracellular adenosine is largely converted to AMP but not ATP. We hypothesized that decreased AMP/ATP ratio underlies the reduced 2,3-BPG production by lowering AMPK activity and subsequently decreasing BPG mutase (BPGM) activity. We measured AMPK phosphorylation and BPGM activity in the RBCs from E1FE and EPO dams. Both AMPK and BPGM activity were decreased in RBCs of E1FE dams compared to controls. Thus, we conclude that i) adenosine derived from uptake via ENT1 is largely converted to AMP; ii) lack of maternal RBC ENT1 lowers AMP/ATP ratio and activity of AMPK and BPGM in maternal RBC. We conducted immunofluorescence staining to assess hypoxia in the placentas, and confirmed the increased expression of HIF-1α in the placentas from E1FE dams. To determine functional changes of these placentas, we conducted metabolomics profiling in both the placenta and maternal plasma. Of all the metabolites altered, amino acids (AA) were the most reduced metabolites in E1FE placentas. In contrast, AA were the most accumulated in maternal plasma. We then injected isotopically labelled 13C15N AA mix in both controls and EIFE dams 24 hours prior to sacrifice. 13C15N AA level was decreased in the placentas in EIFE compared to the controls, while it was accumulated in plasma of EIFE, indicating reduced AA transporter function in the placentas of EIFE. Finally, we performed real time PCR to quantify the mRNA of the known main transporters of AA in the mouse placenta. It showed reduction of LAT1 mRNA in E1FE placenta, where there was no difference in LAT2, SNAT1, or SNAT2. Western blot of the placenta lysates confirmed the expression of LAT1 was indeed reduced. To validate our mouse finding and determine if HIF-1α elevation directly induces LAT1 mRNA in humans, we treated cultured human trophoblast cell line (HTR-8/SVneo cells) with or without DMOG, a cell permeable prolyl-4-hydroxylase inhibitor. After confirming DMOG upregulated HIF-1α, we found stabilized HIF-1α induced LAT1 mRNA levels. Thus, we conclude that elevated HIF-1α underlies the reduction of LAT1 mRNA in cultured human trophoblasts. Conclusion: Our findings suggest that maternal RBCs' oxygen delivering capacity mediated by ENT1 is essential for maintaining adequate placental oxygenation to support fetal growth via AA transporter function. Strategies to improve RBCs' function to deliver oxygen may provide new therapeutic possibilities for FGR. Figure Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document